Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Immunol ; 198(10): 4012-4024, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28416600

ABSTRACT

RNA-based vaccines have recently emerged as a promising alternative to the use of DNA-based and viral vector vaccines, in part because of the potential to simplify how vaccines are made and facilitate a rapid response to newly emerging infections. SAM vaccines are based on engineered self-amplifying mRNA (SAM) replicons encoding an Ag, and formulated with a synthetic delivery system, and they induce broad-based immune responses in preclinical animal models. In our study, in vivo imaging shows that after the immunization, SAM Ag expression has an initial gradual increase. Gene expression profiling in injection-site tissues from mice immunized with SAM-based vaccine revealed an early and robust induction of type I IFN and IFN-stimulated responses at the site of injection, concurrent with the preliminary reduced SAM Ag expression. This SAM vaccine-induced type I IFN response has the potential to provide an adjuvant effect on vaccine potency, or, conversely, it might establish a temporary state that limits the initial SAM-encoded Ag expression. To determine the role of the early type I IFN response, SAM vaccines were evaluated in IFN receptor knockout mice. Our data indicate that minimizing the early type I IFN responses may be a useful strategy to increase primary SAM expression and the resulting vaccine potency. RNA sequence modification, delivery optimization, or concurrent use of appropriate compounds might be some of the strategies to finalize this aim.


Subject(s)
Drug Design , Interferon Type I/immunology , RNA, Messenger/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic , Animals , Antibodies, Viral , Antigens/immunology , Imaging, Three-Dimensional/methods , Interferon Type I/biosynthesis , Mice , RNA, Messenger/administration & dosage , RNA, Messenger/physiology , RNA, Viral/immunology , Respiratory Syncytial Viruses/chemistry , Respiratory Syncytial Viruses/immunology , Vaccination , Vaccine Potency , Viral Vaccines/genetics
2.
J Virol ; 89(4): 2192-200, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25473055

ABSTRACT

UNLABELLED: Although live-attenuated measles virus (MV) vaccines have been used successfully for over 50 years, the target cells that sustain virus replication in vivo are still unknown. We generated a reverse genetics system for the live-attenuated MV vaccine strain Edmonston-Zagreb (EZ), allowing recovery of recombinant (r)MV(EZ). Three recombinant viruses were generated that contained the open reading frame encoding enhanced green fluorescent protein (EGFP) within an additional transcriptional unit (ATU) at various positions within the genome. rMV(EZ)EGFP(1), rMV(EZ)EGFP(3), and rMV(EZ)EGFP(6) contained the ATU upstream of the N gene, following the P gene, and following the H gene, respectively. The viruses were compared in vitro by growth curves, which indicated that rMV(EZ)EGFP(1) was overattenuated. Intratracheal infection of cynomolgus macaques with these recombinant viruses revealed differences in immunogenicity. rMV(EZ)EGFP(1) and rMV(EZ)EGFP(6) did not induce satisfactory serum antibody responses, whereas both in vitro and in vivo rMV(EZ)EGFP(3) was functionally equivalent to the commercial MV(EZ)-containing vaccine. Intramuscular vaccination of macaques with rMV(EZ)EGFP(3) resulted in the identification of EGFP(+) cells in the muscle at days 3, 5, and 7 postvaccination. Phenotypic characterization of these cells demonstrated that muscle cells were not infected and that dendritic cells and macrophages were the predominant target cells of live-attenuated MV. IMPORTANCE: Even though MV strain Edmonston-Zagreb has long been used as a live-attenuated vaccine (LAV) to protect against measles, nothing is known about the primary cells in which the virus replicates in vivo. This is vital information given the push to move toward needle-free routes of vaccination, since vaccine virus replication is essential for vaccination efficacy. We have generated a number of recombinant MV strains expressing enhanced green fluorescent protein. The virus that best mimicked the nonrecombinant vaccine virus was formulated according to protocols for production of commercial vaccine virus batches, and was subsequently used to assess viral tropism in nonhuman primates. The virus primarily replicated in professional antigen-presenting cells, which may explain why this LAV is so immunogenic and efficacious.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , Macrophages/immunology , Macrophages/virology , Measles Vaccine/immunology , Measles virus/immunology , Muscles/immunology , Animals , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Macaca fascicularis , Male , Measles Vaccine/administration & dosage , Measles Vaccine/genetics , Staining and Labeling , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology
3.
PLoS One ; 9(10): e110120, 2014.
Article in English | MEDLINE | ID: mdl-25295727

ABSTRACT

Cotton rats (Sigmodon hispidus) replicate measles virus (MV) after intranasal infection in the respiratory tract and lymphoid tissue. We have cloned the cotton rat signaling lymphocytic activation molecule (CD150, SLAM) in order to investigate its role as a potential receptor for MV. Cotton rat CD150 displays 58% and 78% amino acid homology with human and mouse CD150, respectively. By staining with a newly generated cotton rat CD150 specific monoclonal antibody expression of CD150 was confirmed in cotton rat lymphoid cells and in tissues with a pattern of expression similar to mouse and humans. Previously, binding of MV hemagglutinin has been shown to be dependent on amino acids 60, 61 and 63 in the V region of CD150. The human molecule contains isoleucine, histidine and valine at these positions and binds to MV-H whereas the mouse molecule contains valine, arginine and leucine and does not function as a receptor for MV. In the cotton rat molecule, amino acids 61 and 63 are identical with the mouse molecule and amino acid 60 with the human molecule. After transfection with cotton rat CD150 HEK 293 T cells became susceptible to infection with single cycle VSV pseudotype virus expressing wild type MV glycoproteins and with a MV wildtype virus. After infection, cells expressing cotton rat CD150 replicated virus to lower levels than cells expressing the human molecule and formed smaller plaques. These data might explain why the cotton rat is a semipermissive model for measles virus infection.


Subject(s)
Antigens, CD/metabolism , Measles virus/metabolism , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Antigens, CD/chemistry , Antigens, CD/genetics , Chlorocebus aethiops , Cloning, Molecular , HEK293 Cells , Humans , Measles virus/physiology , Mice , Molecular Sequence Data , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Sigmodontinae , Signaling Lymphocytic Activation Molecule Family Member 1 , Transfection , Vero Cells , Virus Replication
4.
Mol Ther ; 22(12): 2118-2129, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25027661

ABSTRACT

Nucleic acid-based vaccines such as viral vectors, plasmid DNA, and mRNA are being developed as a means to address a number of unmet medical needs that current vaccine technologies have been unable to address. Here, we describe a cationic nanoemulsion (CNE) delivery system developed to deliver a self-amplifying mRNA vaccine. This nonviral delivery system is based on Novartis's proprietary adjuvant MF59, which has an established clinical safety profile and is well tolerated in children, adults, and the elderly. We show that nonviral delivery of a 9 kb self-amplifying mRNA elicits potent immune responses in mice, rats, rabbits, and nonhuman primates comparable to a viral delivery technology, and demonstrate that, relatively low doses (75 µg) induce antibody and T-cell responses in primates. We also show the CNE-delivered self-amplifying mRNA enhances the local immune environment through recruitment of immune cells similar to an MF59 adjuvanted subunit vaccine. Lastly, we show that the site of protein expression within the muscle and magnitude of protein expression is similar to a viral vector. Given the demonstration that self-amplifying mRNA delivered using a CNE is well tolerated and immunogenic in a variety of animal models, we are optimistic about the prospects for this technology.


Subject(s)
Drug Delivery Systems/methods , Emulsions/administration & dosage , Immunity, Cellular , RNA, Messenger/immunology , RNA, Viral/immunology , Vaccines, DNA/administration & dosage , Animals , Cations , Emulsions/chemistry , Female , Macaca mulatta , Mice , Mice, Inbred BALB C , Rabbits , Rats
5.
J Virol ; 87(2): 998-1009, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23135720

ABSTRACT

The major inducible 70-kDa heat shock protein (hsp70) is host protective in a mouse model of measles virus (MeV) brain infection. Transgenic constitutive expression of hsp70 in neurons, the primary target of MeV infection, abrogates neurovirulence in neonatal H-2(d) congenic C57BL/6 mice. A significant level of protection is retained after depletion of T lymphocytes, implicating innate immune mechanisms. The focus of the present work was to elucidate the basis for hsp70-dependent innate immunity using this model. Transcriptome analysis of brains from transgenic (TG) and nontransgenic (NT) mice 5 days after infection identified type I interferon (IFN) signaling, macrophage activation, and antigen presentation as the main differences linked to survival. The pivotal role of type I IFN in hsp70-mediated protection was demonstrated in mice with a genetically disrupted type I IFN receptor (IFNAR(-/-)), where IFNAR(-/-) eliminated the difference in survival between TG and NT mice. Brain macrophages, not neurons, are the predominant source of type I IFN in the virus-infected brain, and in vitro studies provided a mechanistic basis by which MeV-infected neurons can induce IFN-ß in uninfected microglia in an hsp70-dependent manner. MeV infection induced extracellular release of hsp70 from mouse neuronal cells that constitutively express hsp70, and extracellular hsp70 induced IFN-ß transcription in mouse microglial cells through Toll-like receptors 2 and 4. Collectively, our results support a novel axis of type I IFN-dependent antiviral immunity in the virus-infected brain that is driven by hsp70.


Subject(s)
Brain/immunology , HSP70 Heat-Shock Proteins/immunology , Interferon Type I/immunology , Measles virus/immunology , Measles/immunology , Signal Transduction , Animals , Brain/pathology , Brain/virology , Disease Models, Animal , Macrophages/immunology , Male , Measles/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptor, Interferon alpha-beta/deficiency , Survival Analysis , Transcriptome
6.
J Virol ; 85(1): 200-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20962092

ABSTRACT

Measles virus (MV) vaccine effectively protects seronegative individuals against infection. However, inhibition of vaccine-induced seroconversion by maternal antibodies after vaccination remains a problem, as it leaves infants susceptible to MV infection. In cotton rats, passive transfer of MV-specific IgG mimics maternal antibodies and inhibits vaccine-induced seroconversion. Here, we report that immunization in the presence of passively transferred IgG inhibits the secretion of neutralizing antibodies but not the generation of MV-specific B cells. This finding suggested that MV-specific B cells require an additional stimulus to mature into antibody-secreting plasma cells. In order to provide such a stimulus, we generated a recombinant Newcastle disease virus (NDV) expressing the MV hemagglutinin (NDV-H). In contrast to MV, NDV-H induced high levels of type I interferon in plasmacytoid dendritic cells and in lung tissue. In cotton rats immunized with NDV-H, neutralizing antibodies were also generated in the presence of passively transferred antibodies. In the latter case, however, the level and kinetics of antibody generation were reduced. In vitro, alpha interferon stimulated the activation of MV-specific B cells from MV-immune spleen cells. NDV infection (which induces alpha interferon) had the same effect, and stimulation could be abrogated by antibodies neutralizing alpha interferon, but not interleukin 6 (IL-6). In vivo, coapplication of UV-inactivated MV with NDV led to increased MV-specific antibody production in the presence and absence of passively transferred antibodies. These data indicate that MV-specific B cells are being generated after immunization in the presence of maternal antibodies and that the provision of alpha interferon as an additional signal leads to antibody secretion.


Subject(s)
Antibodies, Viral/biosynthesis , B-Lymphocytes/immunology , Hemagglutinins, Viral/immunology , Hemagglutinins, Viral/metabolism , Immunity, Maternally-Acquired/immunology , Interferon Type I/immunology , Measles virus/metabolism , Newcastle disease virus/metabolism , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibodies, Viral/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Female , Hemagglutinins, Viral/genetics , Immunization , Interferon Type I/metabolism , Interferon-alpha/immunology , Measles/immunology , Measles/prevention & control , Measles/virology , Measles virus/genetics , Newcastle disease virus/genetics , Newcastle disease virus/immunology , Recombination, Genetic , Sigmodontinae , Vero Cells
7.
J Virol ; 83(11): 5544-55, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19321604

ABSTRACT

In vitro studies show that hsp70 promotes gene expression for multiple viral families, although there are few reports on the in vivo significance of virus-hsp70 interaction. Previously we showed that hsp70-dependent stimulation of Edmonston measles virus (Ed MeV) transcription caused an increased cytopathic effect and mortality in transgenic hsp70-overexpressing C57BL/6 mice (H-2(b)). The response to MeV infection is influenced by the major histocompatibility complex haplotype; H-2(d) mice are resistant to brain infection due to robust antiviral immune responses, whereas H-2(b) mice are susceptible due to deficiencies in this response. We therefore tested the hypothesis that the outcome of MeV-hsp70 interaction may be dependent upon the host H-2 haplotype. The impact of selective neuronal hsp70 overexpression on Ed MeV brain infection was tested with congenic C57BL/10 H-2(d) neonatal mice. In this context, hsp70 overexpression conferred complete protection against virus-induced mortality, compared to >30% mortality in nontransgenic mice. Selective depletion of T-cell populations showed that transgenic mice exhibit a diminished reliance on T cells for protection. Brain transcript analysis indicated enhanced innate immune activation and signaling through Toll-like receptors 2 and 4 at early times postinfection for transgenic infected mice relative to those for nontransgenic infected mice. Collectively, results suggest that hsp70 can enhance innate antiviral immunity through Toll-like receptor signaling, supporting a protective role for physiological responses that enhance tissue levels of hsp70 (e.g., fever), and that the H-2 haplotype determines the effectiveness of this response.


Subject(s)
Brain Diseases/immunology , HSP72 Heat-Shock Proteins/immunology , HSP72 Heat-Shock Proteins/metabolism , Major Histocompatibility Complex/immunology , Measles virus/immunology , Measles virus/pathogenicity , Measles/immunology , Animals , Brain Diseases/genetics , Brain Diseases/metabolism , Brain Diseases/pathology , Cells, Cultured , Female , Gene Expression Regulation , HSP72 Heat-Shock Proteins/genetics , Haplotypes , Humans , Immunity, Innate/immunology , Male , Measles/genetics , Measles/metabolism , Measles/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Survival Rate , T-Lymphocyte Subsets/immunology , Virulence
8.
Prog Brain Res ; 162: 395-415, 2007.
Article in English | MEDLINE | ID: mdl-17645929

ABSTRACT

Heat shock proteins (HSPs) play an important role in the maintenance of cellular homeostasis, particularly in response to stressful conditions that adversely affect normal cellular structure and function, such as hyperthermia. A remarkable intrinsic resistance of brain to hyperthermia reflects protection mediated by constitutive and induced expression of HSPs in both neurons and glia. Induced expression underlies the phenomenon of hyperthermic pre-reconditioning, where transient, low-intensity heating induces HSPs that protect brain from subsequent insult, reflecting the prolonged half-life of HSPs. The expression and activity of HSPs that is characteristic of nervous tissue plays a role not just in the maintenance and defense of cellular viability, but also in the preservation of neuron-specific luxury functions, particularly those that support synaptic activity. In response to hyperthermia, HSPs mediate preservation or rapid recovery of synaptic function up to the point where damage in other organ systems becomes evident and life threatening. Given the ability of HSPs to enhance gene expression by neurotropic viruses, the constitutive and inducible HSP expression profiles would seem to place nervous tissues at risk. However, we present evidence that the virus-HSP relationship can promote viral clearance in animals capable of mounting effective virus-specific cell-mediated immune responses, potentially reflecting HSP-dependent increases in viral antigenic burden, immune adjuvant effects and cross-presentation of viral antigen. Thus, the protective functions of HSPs span the well-characterized intracellular roles as chaperones to those that may directly or indirectly promote immune function.


Subject(s)
Brain/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Response/physiology , Animals , Fever/physiopathology , Fever/prevention & control , Gene Expression Regulation/physiology , Humans
9.
J Virol ; 80(22): 11031-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16971451

ABSTRACT

Transient hyperthermia such as that experienced during febrile episodes increases expression of the major inducible 70-kDa heat shock protein (hsp72). Despite the relevance of febrile episodes to viral pathogenesis and the multiple in vitro roles of heat shock proteins in viral replication and gene expression, the in vivo significance of virus-heat shock protein interactions is unknown. The present work determined the in vivo relationship between hsp72 levels and neurovirulence of an hsp72-responsive virus using the mouse model of measles virus (MV) encephalitis. Transgenic C57BL/6 mice were created to constitutively overexpress hsp72 in neurons, and these mice were inoculated intracranially with Edmonston MV (Ed MV) at 42 h of age. The mean viral RNA burden in brain was approximately 2 orders of magnitude higher in transgenic animals than in nontransgenic animals 2 to 4 weeks postinfection, and this increased burden was associated with a fivefold increase in mortality. Mice were also challenged with an Ed MV variant exhibiting an attenuated in vitro response to hsp72-dependent stimulation of viral transcription (Ed N-522D). This virus exhibited an attenuated neuropathogenicity in transgenic mice, where mortality and viral RNA burdens were not significantly different from nontransgenic mice infected with either Ed N-522D or parent Ed MV. Collectively, these results indicate that hsp72 levels can serve as a host determinant of viral neurovirulence in C57BL/6 mice, reflecting the direct influence of hsp72 on viral gene expression.


Subject(s)
HSP72 Heat-Shock Proteins/physiology , Measles virus/pathogenicity , Measles/virology , Virulence , Animals , Brain/pathology , Brain/virology , Disease Models, Animal , Encephalitis, Viral/mortality , Encephalitis, Viral/virology , Female , Gene Expression , HSP72 Heat-Shock Proteins/genetics , Histocytochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Viral/analysis , Survival Analysis
10.
J Virol ; 80(6): 2904-12, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16501099

ABSTRACT

The major inducible 70-kDa heat shock protein (hsp72) increases measles virus (MV) transcription and genome replication. This stimulatory effect is attributed to hsp72 interaction with two highly conserved hydrophobic domains in the nucleocapsid protein (N) C terminus of Edmonston MV. These domains are known as Box-2 and Box-3. A single amino acid substitution in Box-3 of Edmonston MV (i.e., N522D) disrupts hsp72 binding. The prevalence of the N522D substitution in contemporary wild-type MV isolates suggests that this sequence has been positively selected. The present work determined if the N522D substitution enhances viral fitness and the degree to which any fitness advantage is influenced by hsp72 levels. Both parent Edmonston MV (Ed N) and an N522D substitution mutant (Ed N-522D) exhibited similar growth on Vero and murine neuroblastoma cells and in cotton rat lung, although Ed N-522D virus exhibited an attenuated in vitro response to hsp72 overexpression. In contrast, mixed infections showed a significantly reduced in vitro and in vivo fitness of Ed N-522D virus. Results support the involvement of additional selectional pressures that maintain the circulation of virus containing N-522D despite the cost to viral fitness.


Subject(s)
Codon , Gene Expression Regulation, Viral , Measles virus/growth & development , Measles virus/pathogenicity , Nucleocapsid Proteins/genetics , Amino Acid Substitution , Animals , Cell Line , Chlorocebus aethiops , HSP72 Heat-Shock Proteins/metabolism , Humans , Lung/virology , Measles/virology , Measles virus/genetics , Mice , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/metabolism , Sigmodontinae , Vero Cells
11.
Virology ; 337(1): 162-74, 2005 Jun 20.
Article in English | MEDLINE | ID: mdl-15914229

ABSTRACT

The major inducible 70-kDa heat shock protein (hsp72) binds measles virus (MV) nucleocapsids and increases MV gene expression. The cytoplasmic tail of the MV N protein (N(TAIL)) contains three hydrophobic domains (Box-1-3) that are potential targets of hsp72 interaction. Low affinity binding to Box-3 is correlated to hsp72-dependent stimulation of MV minireplicon reporter gene expression whereas interactions between hsp72 and Box-1 and/or -2 have not been documented. The present work showed that virus deficient in Box-3/hsp72 interaction retains the ability to form nucleocapsid/hsp72 complexes, identifying Box-2 but not Box-1 as a mediator of high affinity hsp72 binding. Box-2 is the binding site for the viral P protein X domain (XD), where P tethers the viral polymerase to nucleocapsid in support of transcription and genome replication, and competitive inhibition of XD binding to N(TAIL) by hsp72 was shown. Recognition of a common binding site by P and hsp72 represents a potential mechanism for host cell modulation of viral gene expression.


Subject(s)
Heat-Shock Proteins/metabolism , Measles virus/chemistry , Nucleocapsid Proteins/metabolism , Amino Acid Motifs , Binding Sites , Cell Line , HSP72 Heat-Shock Proteins , Humans/metabolism , Nucleocapsid Proteins/chemistry , Protein Binding , Virus Replication
12.
Brain Res ; 1004(1-2): 73-82, 2004 Apr 09.
Article in English | MEDLINE | ID: mdl-15033421

ABSTRACT

Nervous tissue subjected to hyperthermic pre-conditioning is resistance to numerous insults although in vitro, the same treatment can increase gene expression and cytopathic effect of neurotropic paramyxoviruses, including measles virus (MV). The present work determined whether the in vivo relationship between hyperthermic pre-conditioning and MV infection would be to increase neuropathogenicity or, conversely, to promote clearance. Balb/c mice 36 h of age were exposed to a 41 degrees C hyperthermic treatment for 30 min. Intracranial inoculation of mice with Edmonston MV was performed at 6 h following the heat treatment, a time point exhibiting elevated levels of the major inducible 70-kDa heat shock protein in brain, a hallmark of pre-conditioning. Forty-seven percent of the non-heated animals supported a persistent cytopathic infection at 21-day post infection (PI) based upon the quantitative detection of viral RNA in brain using real time RT-PCR. Cytopathic effect in the infected brains was proportionate to viral RNA burden. In contrast, infected stress conditioned mice lacked significant cytopathic effect and clearance was demonstrated in 95% of the animals. Analysis of shorter post-infection intervals showed that levels of viral RNA in brain were equivalent between stress conditioned and non-conditioned mice at 2 and 7 days PI, with clearance being first evident in both groups at 14 days. The temporal onset and progression of clearance was correlated to splenocyte blastogenic responsiveness to purified MV antigen but not the production of MV-specific antibody. Collectively, these results support the hypothesis that stress conditioning enhances the efficacy of cell-mediated immune responses known to mediate viral clearance from brain.


Subject(s)
Brain/metabolism , Disease Models, Animal , Hyperthermia, Induced/methods , Measles virus/metabolism , Measles/metabolism , Animals , Brain/virology , Female , HSP72 Heat-Shock Proteins , Heat-Shock Proteins/biosynthesis , Measles/virology , Measles virus/chemistry , Mice , Mice, Inbred BALB C , Pregnancy
13.
Viral Immunol ; 15(3): 399-416, 2002.
Article in English | MEDLINE | ID: mdl-12479391

ABSTRACT

Heat shock proteins (HSPs) are recognized for their support of protein metabolism. Interaction with viral proteins also enhances the development of innate and adaptive immune responses against the infecting agent. At the level of the infected cell, HSPs are uniquely expressed on the cell surface, where they represent targets of lymphokine activated killer cells. Necrosis of the infected cell releases complexes of HSP and viral protein, which, in turn, binds antigen-presenting cells (APCs). One effect of binding is to stimulate APC maturation and the release of proinflammatory cytokines, an adjuvant effect that prepares the way for adaptive immune responses. A second effect of binding is to direct the antigenic cargo of the HSP into endogenous MHC presentation pathways for priming of naive cytotoxic T cells (CTL) or activation of antigen-specific CTLs. This alternate pathway of antigen presentation is essential to CTL priming following primary brain infection. Using heat shock to elevate brain levels of HSP in a mouse model of measles virus (MV) persistent infection, we provide evidence supporting a role for HSPs in promoting cell-mediated viral clearance from brain. The findings highlight the probable relevance of HSPs to anti-MV immunity, suggesting novel routes of both therapeutic intervention and preventative measures.


Subject(s)
Heat-Shock Proteins/physiology , Measles virus/immunology , Measles/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Heat-Shock Proteins/immunology , Immunity, Innate , Lymphocyte Activation , Measles/prevention & control , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...